Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Cell ; 81(5): 953-968.e9, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33503407

RESUMEN

While the role of transcription factors and coactivators in controlling enhancer activity and chromatin structure linked to gene expression is well established, the involvement of corepressors is not. Using inflammatory macrophage activation as a model, we investigate here a corepressor complex containing GPS2 and SMRT both genome-wide and at the Ccl2 locus, encoding the chemokine CCL2 (MCP-1). We report that corepressors co-occupy candidate enhancers along with the coactivators CBP (H3K27 acetylase) and MED1 (mediator) but act antagonistically by repressing eRNA transcription-coupled H3K27 acetylation. Genome editing, transcriptional interference, and cistrome analysis reveals that apparently related enhancer and silencer elements control Ccl2 transcription in opposite ways. 4C-seq indicates that corepressor depletion or inflammatory signaling functions mechanistically similarly to trigger enhancer activation. In ob/ob mice, adipose tissue macrophage-selective depletion of the Ccl2 enhancer-transcribed eRNA reduces metaflammation. Thus, the identified corepressor-eRNA-chemokine pathway operates in vivo and suggests therapeutic opportunities by targeting eRNAs in immuno-metabolic diseases.


Asunto(s)
Quimiocina CCL2/genética , Proteínas Co-Represoras/genética , Elementos de Facilitación Genéticos , Péptidos y Proteínas de Señalización Intracelular/genética , Co-Represor 2 de Receptor Nuclear/genética , Obesidad/genética , Elementos Silenciadores Transcripcionales , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Animales , Sistemas CRISPR-Cas , Quimiocina CCL2/inmunología , Proteínas Co-Represoras/inmunología , Edición Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Histonas/genética , Histonas/inmunología , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Masculino , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/inmunología , Ratones , Ratones Obesos , Co-Represor 2 de Receptor Nuclear/inmunología , Obesidad/inmunología , Obesidad/patología , Células RAW 264.7 , ARN no Traducido/genética , ARN no Traducido/inmunología , Transducción de Señal
2.
Nat Immunol ; 17(5): 505-513, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26998764

RESUMEN

The effect of alterations in intestinal microbiota on microbial metabolites and on disease processes such as graft-versus-host disease (GVHD) is not known. Here we carried out an unbiased analysis to identify previously unidentified alterations in gastrointestinal microbiota-derived short-chain fatty acids (SCFAs) after allogeneic bone marrow transplant (allo-BMT). Alterations in the amount of only one SCFA, butyrate, were observed only in the intestinal tissue. The reduced butyrate in CD326(+) intestinal epithelial cells (IECs) after allo-BMT resulted in decreased histone acetylation, which was restored after local administration of exogenous butyrate. Butyrate restoration improved IEC junctional integrity, decreased apoptosis and mitigated GVHD. Furthermore, alteration of the indigenous microbiota with 17 rationally selected strains of high butyrate-producing Clostridia also decreased GVHD. These data demonstrate a heretofore unrecognized role of microbial metabolites and suggest that local and specific alteration of microbial metabolites has direct salutary effects on GVHD target tissues and can mitigate disease severity.


Asunto(s)
Células Epiteliales/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedad Injerto contra Huésped/inmunología , Intestinos/inmunología , Metaboloma/inmunología , Acetilación/efectos de los fármacos , Animales , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/métodos , Butiratos/inmunología , Butiratos/metabolismo , Butiratos/farmacología , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Ácidos Grasos Volátiles/inmunología , Ácidos Grasos Volátiles/metabolismo , Femenino , Cromatografía de Gases y Espectrometría de Masas , Microbioma Gastrointestinal/fisiología , Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/microbiología , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/inmunología , Histona Desacetilasas/metabolismo , Histonas/inmunología , Histonas/metabolismo , Immunoblotting , Intestinos/citología , Intestinos/microbiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
3.
Nat Immunol ; 16(10): 1094-103, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26301565

RESUMEN

B lymphopoiesis requires that immunoglobulin genes be accessible to RAG1-RAG2 recombinase. However, the RAG proteins bind widely to open chromatin, which suggests that additional mechanisms must restrict RAG-mediated DNA cleavage. Here we show that developmental downregulation of interleukin 7 (IL-7)-receptor signaling in small pre-B cells induced expression of the bromodomain-family member BRWD1, which was recruited to a specific epigenetic landscape at Igk dictated by pre-B cell receptor (pre-BCR)-dependent Erk activation. BRWD1 enhanced RAG recruitment, increased gene accessibility and positioned nucleosomes 5' to each Jκ recombination signal sequence. BRWD1 thus targets recombination to Igk and places recombination within the context of signaling cascades that control B cell development. Our findings represent a paradigm in which, at any particular antigen-receptor locus, specialized mechanisms enforce lineage- and stage-specific recombination.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Inmunoglobulinas/genética , Recombinación Genética/inmunología , Animales , Apoptosis , Regulación hacia Abajo/inmunología , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Interleucina-7/genética , Interleucina-7/inmunología , Ratones , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología
4.
J Immunol ; 206(5): 1077-1087, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33483347

RESUMEN

The activation of T cells is accompanied by intensive posttranscriptional remodeling of their proteome. We observed that protein expression of enzymes that modify wobble uridine in specific tRNAs, namely elongator subunit 3 (Elp3) and cytosolic thiouridylase (Ctu)2, increased in the course of T cell activation. To investigate the role of these tRNA epitranscriptomic modifiers in T cell biology, we generated mice deficient for Elp3 in T cells. We show that deletion of Elp3 has discrete effects on T cells. In vitro, Elp3-deficient naive CD4+ T cells polarize normally but are delayed in entering the first cell cycle following activation. In vivo, different models of immunization revealed that Elp3-deficient T cells display reduced expansion, resulting in functional impairment of T follicular helper (TFH) responses, but not of other CD4+ effector T cell responses. Transcriptomic analyses identified a progressive overactivation of the stress-responsive transcription factor Atf4 in Elp3-deficient T cells. Overexpression of Atf4 in wild-type T cells phenocopies the effect of Elp3 loss on T cell cycle entry and TFH cell responses. Reciprocally, partial silencing of Atf4 or deletion of its downstream effector transcription factor Chop rescues TFH responses of Elp3-deficient T cells. Together, our results reveal that specific epitranscriptomic tRNA modifications contribute to T cell cycle entry and promote optimal TFH responses.


Asunto(s)
Factor de Transcripción Activador 4/genética , Histona Acetiltransferasas/genética , ARN de Transferencia/genética , Células T Auxiliares Foliculares/inmunología , Uridina/genética , Factor de Transcripción Activador 4/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Femenino , Histona Acetiltransferasas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Procesamiento Postranscripcional del ARN/genética , Procesamiento Postranscripcional del ARN/inmunología , ARN de Transferencia/inmunología , Transcriptoma/genética , Transcriptoma/inmunología , Uridina/inmunología
5.
Immunity ; 36(5): 717-30, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22579476

RESUMEN

Regulatory T (Treg) cells, driven by the Foxp3 transcription factor, are responsible for limiting autoimmunity and chronic inflammation. We showed that a well-characterized Foxp3(gfp) reporter mouse, which expresses an N-terminal GFP-Foxp3 fusion protein, is a hypomorph that causes profoundly accelerated autoimmune diabetes on a NOD background. Although natural Treg cell development and in vitro function are not markedly altered in Foxp3(gfp) NOD and C57BL/6 mice, Treg cell function in inflammatory environments was perturbed and TGF-ß-induced Treg cell development was reduced. Foxp3(gfp) was unable to interact with the histone acetyltransferase Tip60, the histone deacetylase HDAC7, and the Ikaros family zinc finger 4, Eos, which led to reduced Foxp3 acetylation and enhanced K48-linked polyubiquitylation. Collectively this results in an altered transcriptional landscape and reduced Foxp3-mediated gene repression, notably at the hallmark IL-2 promoter. Loss of controlled Foxp3-driven epigenetic modification leads to Treg cell insufficiency that enables autoimmunity in susceptible environments.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/inmunología , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Acetilación , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Epigénesis Genética , Factores de Transcripción Forkhead/genética , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/inmunología , Histona Desacetilasas/metabolismo , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-2/metabolismo , Lisina Acetiltransferasa 5 , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas , Linfocitos T Reguladores/metabolismo , Transactivadores/genética , Transactivadores/inmunología , Transactivadores/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo
6.
FASEB J ; 32(10): 5312-5325, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29718706

RESUMEN

A key event required for effective resolution of inflammation is efferocytosis, which is defined as phagocytic removal of apoptotic cells mostly by macrophages acquiring an alternatively activated phenotype (M2). c-Myc has been reported to play a role in alternative activation of human macrophages and is proposed as one of the M2 macrophage markers. We found that M2-like peritoneal macrophages from zymosan A-treated mice exhibited a marked accumulation of Myc-nick, a truncated protein generated by a Calpain-mediated proteolytic cleavage of full-length c-Myc. Further, ectopic expression of Myc-nick in murine bone marrow-derived macrophages promoted the M2 polarization and, consequently, enhanced their efferocytic capability. Notably, Myc-nick-induced efferocytosis was found to be tightly associated with α-tubulin acetylation by K acetyltransferase 2a (Kat2a/Gcn5) activity. These findings suggest Myc-nick as a novel proresolving mediator that has a fundamental function in maintaining homeostasis under inflammatory conditions.-Zhong, X., Lee, H.-N., Kim, S. H., Park, S.-A., Kim, W., Cha, Y.-N., Surh, Y.-J. Myc-nick promotes efferocytosis through M2 macrophage polarization during resolution of inflammation.


Asunto(s)
Células de la Médula Ósea/inmunología , Macrófagos Peritoneales/inmunología , Proteínas Proto-Oncogénicas c-myc/inmunología , Acetilación , Animales , Antígenos de Diferenciación/inmunología , Células de la Médula Ósea/patología , Histona Acetiltransferasas/inmunología , Inflamación/inmunología , Inflamación/patología , Macrófagos Peritoneales/patología , Ratones , Tubulina (Proteína)/inmunología , Factores de Transcripción p300-CBP/inmunología
7.
Blood ; 125(8): 1325-32, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25540192

RESUMEN

Circulating thrombin-activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) are causal factors for thrombolytic failure. Therefore, we evaluated an antibody-engineered bispecific inhibitor against TAFI and PAI-1 (heterodimer diabody, Db-TCK26D6x33H1F7) in several mouse models of thrombosis and stroke. Prophylactic administration of the diabody (0.8 mg/kg) in a thromboplastin-induced model of thromboembolism led to decreased lung fibrin deposition. In a model of cerebral ischemia and reperfusion, diabody administration (0.8 mg/kg, 1 hour postocclusion) led to a mitigated cerebral injury with a 2.3-fold reduced lesion and improved functional outcomes. In a mouse model of thrombin-induced middle cerebral artery occlusion, the efficacy of the diabody was compared to the standard thrombolytic treatment with recombinant tissue-type plasminogen activator (tPA). Early administration of diabody (0.8 mg/kg) caused a twofold decrease in brain lesion size, whereas that of tPA (10 mg/kg) had a much smaller effect. Delayed administration of diabody or tPA had no effect on lesion size, whereas the combined administration of diabody with tPA caused a 1.7-fold decrease in lesion size. In contrast to tPA, the diabody did not increase accumulative bleeding. In conclusion, administration of a bispecific inhibitor against TAFI and PAI-1 results in a prominent profibrinolytic effect in mice without increased bleeding.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Fibrinolíticos/uso terapéutico , Histona Acetiltransferasas/inmunología , Serpina E2/inmunología , Accidente Cerebrovascular/terapia , Factores Asociados con la Proteína de Unión a TATA/inmunología , Terapias en Investigación/métodos , Factor de Transcripción TFIID/inmunología , Tromboembolia Venosa/terapia , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos C57BL , Multimerización de Proteína , Accidente Cerebrovascular/patología , Tromboembolia Venosa/patología
8.
Proc Natl Acad Sci U S A ; 111(26): 9585-90, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24979783

RESUMEN

Memory B cells and long-lived bone marrow-resident plasma cells maintain humoral immunity. Little is known about the intrinsic mechanisms that are essential for forming memory B cells or endowing them with the ability to rapidly differentiate upon reexposure while maintaining the population over time. Histone modifications have been shown to regulate lymphocyte development, but their role in regulating differentiation and maintenance of B-cell subsets during an immune response is unclear. Using stage-specific deletion of monocytic leukemia zinc finger protein (MOZ), a histone acetyltransferase, we demonstrate that mutation of this chromatin modifier alters fate decisions in both primary and secondary responses. In the absence of MOZ, germinal center B cells were significantly impaired in their ability to generate dark zone centroblasts, with a concomitant decrease in both cell-cycle progression and BCL-6 expression. In contrast, there was increased differentiation to IgM and low-affinity IgG1(+) memory B cells. The lack of MOZ affected the functional outcome of humoral immune responses, with an increase in secondary germinal centers and a corresponding decrease in secondary high-affinity antibody-secreting cell formation. Therefore, these data provide strong evidence that manipulating epigenetic modifiers can regulate fate decisions during humoral responses, and thus could be targeted for therapeutic intervention.


Asunto(s)
Linfocitos B/inmunología , Regulación de la Expresión Génica/inmunología , Centro Germinal/inmunología , Histona Acetiltransferasas/inmunología , Memoria Inmunológica/inmunología , Animales , Secuencia de Bases , Cromatina/inmunología , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Ratones , Análisis por Micromatrices , Datos de Secuencia Molecular , Análisis de Secuencia de ARN , Estadísticas no Paramétricas
9.
Plant Cell ; 25(2): 762-76, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23435660

RESUMEN

The Arabidopsis thaliana Elongator complex subunit2 (ELP2) genetically interacts with NONEXPRESSOR OF PATHOGENESIS-RELATED GENES1 (NPR1), a key transcription coactivator of plant immunity, and regulates the induction kinetics of defense genes. However, the mechanistic relationship between ELP2 and NPR1 and how ELP2 regulates the kinetics of defense gene induction are unclear. Here, we demonstrate that ELP2 is an epigenetic regulator required for pathogen-induced rapid transcriptome reprogramming. We show that ELP2 functions in a transcriptional feed-forward loop regulating both NPR1 and its target genes. An elp2 mutation increases the total methylcytosine number, reduces the average methylation levels of methylcytosines, and alters (increases or decreases) methylation levels of specific methylcytosines. Interestingly, infection of plants with the avirulent bacterial pathogen Pseudomonas syringae pv tomato DC3000/avrRpt2 induces biphasic changes in DNA methylation levels of NPR1 and PHYTOALEXIN DEFICIENT4 (PAD4), which encodes another key regulator of plant immunity. These dynamic changes are blocked by the elp2 mutation, which is correlated with delayed induction of NPR1 and PAD4. The elp2 mutation also reduces basal histone acetylation levels in the coding regions of several defense genes. Together, our data demonstrate a new role for Elongator in somatic DNA demethylation/methylation and suggest a function for Elongator-mediated chromatin regulation in pathogen-induced transcriptome reprogramming.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Arabidopsis/inmunología , Epigénesis Genética , Histona Acetiltransferasas/metabolismo , Inmunidad de la Planta/genética , Acetilación , Arabidopsis/microbiología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/inmunología , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Metilación de ADN , Epigénesis Genética/inmunología , Regulación de la Expresión Génica de las Plantas , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Histonas/metabolismo , Mutación , Plantas Modificadas Genéticamente , Pseudomonas syringae/patogenicidad
10.
J Immunol ; 192(11): 5285-95, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24778442

RESUMEN

Hyaluronidase (Hyal) and low m.w. hyaluronan (LMW HA) fragments have been widely reported to stimulate the innate immune response. However, most hyaluronidases used were purified from animal tissues (e.g., bovine testis Hyal [BTH]), and contain endotoxin and other unrelated proteins. We tested a highly purified recombinant human Hyal (rHuPH20) and endotoxin-free HA fragments from M(r) 5,000 to 1,500,000 in the rodent air pouch model of inflammation to determine their potential for stimulation of the innate immune response. Exogenous LMW HA fragments (average M(r) 200,000) failed to induce either cytokine/chemokine production or neutrophil infiltration into the air pouch. Challenging the air pouch with LPS or BTH stimulated production of cytokines and chemokines but rHuPH20 did not, suggesting that neither PH20 nor generation of LMW HA fragments in situ stimulates cytokine and chemokine production. LPS and BTH also induced neutrophil infiltration into the air pouch, which was not observed with rHuPH20 treatment. Endotoxin-depleted BTH had much reduced proinflammatory activity, suggesting that the difference in inflammatory responses between rHuPH20 and BTH is likely due to endotoxin contaminants in BTH. When rHuPH20 was dosed with LPS, the induction of cytokines and chemokines was the same as LPS alone, but neutrophil infiltration was inhibited, likely by interrupting HA-CD44 interaction. Our results indicate that neither rHuPH20 nor its directly generated HA catabolites have inflammatory properties in the air pouch model, and rHuPH20 can instead inhibit some aspects of inflammation, such as neutrophil infiltration into the air pouch.


Asunto(s)
Antígenos de Neoplasias/farmacología , Histona Acetiltransferasas/farmacología , Ácido Hialurónico/inmunología , Hialuronoglucosaminidasa/farmacología , Lipopolisacáridos/toxicidad , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Enfermedad Aguda , Animales , Antígenos de Neoplasias/inmunología , Bovinos , Línea Celular , Citocinas/inmunología , Histona Acetiltransferasas/inmunología , Humanos , Hialuronoglucosaminidasa/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Infiltración Neutrófila/inmunología , Neutrófilos/patología , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología
11.
Proc Natl Acad Sci U S A ; 110(26): 10717-22, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23749869

RESUMEN

The Drosophila defense against pathogens largely relies on the activation of two signaling pathways: immune deficiency (IMD) and Toll. The IMD pathway is triggered mainly by Gram-negative bacteria, whereas the Toll pathway responds predominantly to Gram-positive bacteria and fungi. The activation of these pathways leads to the rapid induction of numerous NF-κB-induced immune response genes, including antimicrobial peptide genes. The IMD pathway shows significant similarities with the TNF receptor pathway. Recent evidence indicates that the IMD pathway is also activated in response to various noninfectious stimuli (i.e., inflammatory-like reactions). To gain a better understanding of the molecular machinery underlying the pleiotropic functions of this pathway, we first performed a comprehensive proteomics analysis to identify the proteins interacting with the 11 canonical members of the pathway initially identified by genetic studies. We identified 369 interacting proteins (corresponding to 291 genes) in heat-killed Escherichia coli-stimulated Drosophila S2 cells, 92% of which have human orthologs. A comparative analysis of gene ontology from fly or human gene annotation databases points to four significant common categories: (i) the NuA4, nucleosome acetyltransferase of H4, histone acetyltransferase complex, (ii) the switching defective/sucrose nonfermenting-type chromatin remodeling complex, (iii) transcription coactivator activity, and (iv) translation factor activity. Here we demonstrate that sumoylation of the IκB kinase homolog immune response-deficient 5 plays an important role in the induction of antimicrobial peptide genes through a highly conserved sumoylation consensus site during bacterial challenge. Taken together, the proteomics data presented here provide a unique avenue for a comparative functional analysis of proteins involved in innate immune reactions in flies and mammals.


Asunto(s)
Proteínas de Drosophila/inmunología , Drosophila/inmunología , Drosophila/microbiología , Transducción de Señal/inmunología , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Escherichia coli/inmunología , Genes de Insecto , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Histona Acetiltransferasas/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mapas de Interacción de Proteínas , Homología de Secuencia de Aminoácido
12.
J Biol Chem ; 288(22): 15537-46, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23609452

RESUMEN

The abundant expression of IFNγ in Th-inducing POK (ThPOK)-deficient CD4(+) T cells requires the activation of Eomesodermin (Eomes); however, the underlying mechanism of this phenomenon remains unclear. Here we report that ThPOK binds directly to the promoter region of the Eomes gene to repress its expression in CD4(+) T cells. We identified the histone acetyltransferase TIP60 as a co-repressor of ThPOK-target genes, where ectopically expressed TIP60 increased ThPOK protein stability by promoting its acetylation at its Lys(360) residue to then augment the transcriptional repression of Eomes. Moreover, knockdown of endogenous TIP60 abolished the stabilization of ThPOK in CD4(+) T cells, which led to the transcriptional activation of Eomes and increased production of IFNγ. Our results reveal a novel pathway by which TIP60 and ThPOK synergistically suppresses Eomes function and IFNγ production, which could contribute to the regulation of inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/inmunología , Histona Acetiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica/genética , Células HEK293 , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Lisina Acetiltransferasa 5 , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Estabilidad Proteica , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
13.
Immunology ; 139(2): 161-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23347099

RESUMEN

The MOnocytic leukaemia Zing finger (MOZ; MYST3 or KAT6A(1)) gene is frequently found translocated in acute myeloid leukaemia. MOZ encodes a large multidomain protein that contains, besides others, a histone acetyl transferase catalytic domain. Several studies have now established the critical function of MOZ in haematopoiesis. In this review we summarize the recent findings that underscore the relevance of the different biological activities of MOZ in the regulation of haematopoiesis.


Asunto(s)
Epigénesis Genética , Hematopoyesis/genética , Histona Acetiltransferasas/genética , Transactivadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Histona Acetiltransferasas/inmunología , Histona Acetiltransferasas/metabolismo , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Modelos Genéticos , Modelos Inmunológicos
14.
J Autoimmun ; 47: 34-44, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24012345

RESUMEN

Maintenance of FOXP3 protein expression is crucial for differentiation and maturation of regulatory T (Treg) cells, which play important roles in immune homeostasis and immune tolerance. We demonstrate here that PDCD5 interacts with FOXP3, increases acetylation of FOXP3 in synergy with Tip60 and enhances the repressive function of FOXP3. In PDCD5 transgenic (PDCD5tg) mice, overexpression of PDCD5 enhanced the level of FOXP3 protein and percentage of CD4(+)CD25(+)FOXP3(+) cells. Naïve CD4(+) T cells from PDCD5tg mice were more sensitive to TGF-ß-induced Treg polarization and expansion. These induced Tregs retained normal suppressive function in vitro. Severity of experimentally-induced autoimmune encephalomyelitis (EAE) in PDCD5tg mice was significantly reduced relative to that of wild-type mice. The beneficial effect of PDCD5 likely resulted from increases of Treg cell frequency, accompanied by a reduction of the predominant pathogenic Th17/Th1 response. Activation-induced cell death enhanced by PDCD5 was also linked to this process. This is the first report revealing that PDCD5 activity in T cells suppresses autoimmunity by modulating Tregs. This study suggests that PDCD5 serves as a guardian of immunological functions and that the PDCD5-FOXP3-Treg axis may be a therapeutic target for autoimmunity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Autoinmunidad/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Acetilación , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Antígenos CD4/inmunología , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Lisina Acetiltransferasa 5 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño , Transactivadores/genética , Transactivadores/inmunología , Factor de Crecimiento Transformador beta/inmunología
15.
J Immunol ; 185(10): 6003-12, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20935208

RESUMEN

Recent evidence indicates a new role for histone deacetylases (HDACs) in the activation of genes governing the host immune response. Virus, along with other pathogenic stimuli, triggers an antiviral defense mechanism through the induction of IFN, IFN-stimulated genes, and other proinflammatory cytokines. Many of these genes have been shown to be regulated by transcription factors of the IFN regulatory factor (IRF) family. Recent studies from IRF5 knockout mice have confirmed a critical role for IRF5 in virus-induced type I IFN expression and proinflammatory cytokines IL-6, IL-12, and TNF-α; yet, little is known of the molecular mechanism of IRF5-mediated proinflammatory cytokine expression. In this study, we show that both HDACs and histone acetyltransferases (HATs) associate with IRF5, leading to alterations in its transactivation ability. Using the HDAC inhibitor trichostatin A, we demonstrate that ISRE, IFNA, and IL6 promoters require HDAC activity for transactivation and transcription, whereas TNFα does not. Mapping the interaction of corepressor proteins (HDAC1, silencing mediator of retinoid and thyroid receptor/nuclear corepressor of retinoid receptor, and Sin3a) and HATs to IRF5 revealed distinct differences, including the dependence of IRF5 phosphorylation on HAT association resulting in IRF5 acetylation. Data presented in this study support a mechanism whereby virus triggers the dynamic conversion of an IRF5-mediated silencing complex to that of an activating complex on promoters of target genes. These data provide the first evidence, to our knowledge, of a tightly controlled transcriptional mechanism whereby IRF5 regulates proinflammatory cytokine expression in conjunction with HATs and HDACs.


Asunto(s)
Citocinas/biosíntesis , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Factores Reguladores del Interferón/biosíntesis , Activación Transcripcional/inmunología , Línea Celular , Inmunoprecipitación de Cromatina , Citocinas/inmunología , Expresión Génica , Histona Acetiltransferasas/inmunología , Histona Desacetilasas/inmunología , Humanos , Immunoblotting , Inmunoprecipitación , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Reacción en Cadena de la Polimerasa , Activación Transcripcional/genética , Transfección
16.
Sci Immunol ; 7(67): eabb6032, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35061506

RESUMEN

The autoimmune regulator (AIRE) induces the transcription of thousands of peripheral tissue genes (PTGs) in thymic epithelial cells (TECs) to mediate immunological tolerance. The chromatin state required for optimal AIRE function in TECs and how this state is induced remains unclear. We tested the role of the histone acetyltransferase, KAT7 (also known as HBO1 or MYST2), which is essential for acetylation of histone 3 lysine 14, in TEC differentiation, AIRE-mediated PTG expression, and thymic tolerance. We find that KAT7 is required for optimal expansion of medullary TEC and has a major role in the expression of AIRE-dependent PTGs, associated with enhanced chromatin accessibility at these gene loci in TECs. Mice with TEC-specific Kat7 deletion develop organ-specific autoimmunity with features resembling those observed in Aire-deficient mice. These findings highlight critical roles for KAT7-mediated acetylation in promoting a chromatin state at PTG loci that enables AIRE function and the establishment of immunological tolerance.


Asunto(s)
Células Epiteliales/inmunología , Histona Acetiltransferasas/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Tolerancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Timo/citología , Proteína AIRE
17.
J Immunol ; 183(10): 6422-31, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19880447

RESUMEN

The pool of hematopoietic stem/progenitor cells, which provide life-long reconstitution of all hematopoietic lineages, is tightly controlled and regulated by self-renewal and apoptosis. Histone modifiers and chromatin states are believed to govern establishment, maintenance, and propagation of distinct patterns of gene expression in stem cells, however the underlying mechanism remains poorly understood. In this study, we identified a role for the histone acetytransferase cofactor Trrap in the maintenance of hematopietic stem/progenitor cells. Conditional deletion of the Trrap gene in mice resulted in ablation of bone marrow and increased lethality. This was due to the depletion of early hematopoietic progenitors, including hematopoietic stem cells, via a cell-autonomous mechanism. Analysis of purified bone marrow progenitors revealed that these defects are associated with induction of p53-independent apoptosis and deregulation of Myc transcription factors. Together, this study has identified a critical role for Trrap in the mechanism that maintains hematopoietic stem cells and hematopoietic system, and underscores the importance of Trrap and histone modifications in tissue homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Coenzimas/inmunología , Células Madre Hematopoyéticas/inmunología , Histona Acetiltransferasas/inmunología , Proteínas Nucleares/inmunología , Proteína p53 Supresora de Tumor/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/inmunología , Médula Ósea/inmunología , Médula Ósea/metabolismo , Coenzimas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Histona Acetiltransferasas/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(5): 721-4, 2011 Sep.
Artículo en Zh | MEDLINE | ID: mdl-22007507

RESUMEN

OBJECTIVE: To prepare and identify the monoclonal antibody (mAb) against hMOF. METHODS: BALB/C mice were immunized with protein from the spleen cells isolated and fused with SP2/0 cells. After several rounds of screening and cloning, the hybridoma cell strain secreting anti-hMOF mAb was obtained. Its specificity was evaluated with ELISA and Western blot, and the titer, immunoglobulin subtype and affinity of the mAb were measured. RESULTS: One cell strains of hybridoma were obtained and named as 4C1C8. The anti-hMOF mAb secreted by the hybridoma cell strain was identified as IgG1 subtype. The mAb titers in ascitic fluid were 1 409600, as determined with ELISA with an affinity reaching to 7.65 x 10(6) L/mol. Western blot demonstrated that the antibodies could specifically recognize the immunogen. The cell immunohistochemistry proved that the antibody could recognize the hMOF antigens expressed on the normal cells HL7702. CONCLUSION: The success in anti-hMOF mAb preparation provides the basis for further study of hMOF.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Histona Acetiltransferasas/inmunología , Hibridomas/metabolismo , Animales , Anticuerpos Monoclonales/genética , Femenino , Histona Acetiltransferasas/biosíntesis , Histona Acetiltransferasas/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión
19.
Front Immunol ; 12: 808159, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046960

RESUMEN

Post-translational modifications (PTMs), such as phosphorylation and ubiquitination, etc., have been reported to modulate the activities of IRF3 and IRF7. In this study, we found an acetyltransferase KAT8 in grass carp (CiKAT8, MW286472) that acetylated IRF3/IRF7 and then resulted in inhibition of IFN 1 response. CiKAT8 expression was up-regulated in the cells under poly I:C, B-DNA or Z-DNA stimulation as well as GCRV(strain 873) or SVCV infection. The acetyltransferase domain (MYST domain) of KAT8 promoted the acetylation of IRF3 and IRF7 through the direct interaction with them. So, the domain is essential for KAT8 function. Expectedly, KAT8 without MYST domain (KAT8-△264-487) was granularly aggregated in the nucleus and failed to down-regulate IFN 1 expression. Subcellular localization analysis showed that KAT8 protein was evenly distributed in the nucleus. In addition, we found that KAT8 inhibited the recruitment of IRF3 and IRF7 to ISRE response element. Taken together, our findings revealed that grass carp KAT8 blocked the activities of IRF3 and IRF7 by acetylating them, resulting in a low affinity interaction of ISRE response element with IRF3 and IRF7, and then inhibiting nucleic acids-induced innate immune response.


Asunto(s)
Proteínas de Peces/inmunología , Regulación de la Expresión Génica/inmunología , Histona Acetiltransferasas/inmunología , Inmunidad Innata/inmunología , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/inmunología , Acetilación , Animales , Carpas
20.
Immunol Res ; 40(2): 164-78, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18213528

RESUMEN

Epigenetic modifications of chromatin, such as histone acetylation, are involved in repression of tumor antigens and multiple immune genes that are thought to facilitate tumor escape. The status of acetylation in a cell is determined by the balance of the activities of histone acetyltransferases and histone deacetylases. Inhibitors of histone deacetylase (HDACi) can enhance the expression of immunologically important molecules in tumor cells and HDACi treated tumor cells are able to induce immune responses in vitro and in vivo. Systemic HDACi are in clinical trails in cancer and also being used in several autoimmune disease models. To date, 18 HDACs have been reported in human cells and more than thirty HDACi identified, although only a few immune targets of these inhibitors have been identified. Here, we discuss the molecular pathways employed by HDACi and their potential role in inducing immune responses against tumors. We review data suggesting that selection of target specific HDACi and combinations with other agents and modalities, including those that activate stress pathways, may further enhance the efficacy of epigenetic therapies.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Histona Acetiltransferasas/inmunología , Histona Desacetilasas/inmunología , Histonas/metabolismo , Neoplasias/enzimología , Neoplasias/inmunología , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Cromatina/química , Cromatina/efectos de los fármacos , Ensayos Clínicos como Asunto , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/inmunología , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/química , Humanos , Neoplasias/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA